Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Nat Immunol ; 23(2): 287-302, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35105987

RESUMO

The volume-regulated anion channel (VRAC) is formed by LRRC8 proteins and is responsible for the regulatory volume decrease (RVD) after hypotonic cell swelling. Besides chloride, VRAC transports other molecules, for example, immunomodulatory cyclic dinucleotides (CDNs) including 2'3'cGAMP. Here, we identify LRRC8C as a critical component of VRAC in T cells, where its deletion abolishes VRAC currents and RVD. T cells of Lrrc8c-/- mice have increased cell cycle progression, proliferation, survival, Ca2+ influx and cytokine production-a phenotype associated with downmodulation of p53 signaling. Mechanistically, LRRC8C mediates the transport of 2'3'cGAMP in T cells, resulting in STING and p53 activation. Inhibition of STING recapitulates the phenotype of LRRC8C-deficient T cells, whereas overexpression of p53 inhibits their enhanced T cell function. Lrrc8c-/- mice have exacerbated T cell-dependent immune responses, including immunity to influenza A virus infection and experimental autoimmune encephalomyelitis. Our results identify cGAMP uptake through LRRC8C and STING-p53 signaling as a new inhibitory signaling pathway in T cells and adaptive immunity.


Assuntos
Ânions/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Canais Iônicos/metabolismo , Proteínas de Membrana/metabolismo , Linfócitos T/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Cálcio/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Nucleotídeos Cíclicos/metabolismo , Transdução de Sinais/fisiologia
2.
Am J Physiol Cell Physiol ; 324(5): C1017-C1027, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36878847

RESUMO

Sirtuins are NAD+-dependent deacetylases with beneficial roles in conditions relevant to human health, including metabolic disease, type II diabetes, obesity, cancer, aging, neurodegenerative diseases, and cardiac ischemia. Since ATP-sensitive K+ (KATP) channels have cardioprotective roles, we investigated whether they are regulated by sirtuins. Nicotinamide mononucleotide (NMN) was used to increase cytosolic NAD+ levels and to activate sirtuins in cell lines, isolated rat and mouse cardiomyocytes or insulin-secreting INS-1 cells. KATP channels were studied with patch clamping, biochemistry techniques, and antibody uptake experiments. NMN led to an increase in intracellular NAD+ levels and an increase in the KATP channel current, without significant changes in the unitary current amplitude or open probability. An increased surface expression was confirmed using surface biotinylation approaches. The rate of KATP channel internalization was diminished by NMN, which may be a partial explanation for the increased surface expression. We show that NMN acts via sirtuins since the increased KATP channel surface expression was prevented by blockers of SIRT1 and SIRT2 (Ex527 and AGK2) and mimicked by SIRT1 activation (SRT1720). The pathophysiological relevance of this finding was studied using a cardioprotection assay with isolated ventricular myocytes, in which NMN protected against simulated ischemia or hypoxia in a KATP channel-dependent manner. Overall, our data draw a link between intracellular NAD+, sirtuin activation, KATP channel surface expression, and cardiac protection against ischemic damage.


Assuntos
Diabetes Mellitus Tipo 2 , Sirtuínas , Ratos , Camundongos , Humanos , Animais , Sirtuína 1/genética , Sirtuína 1/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo , NAD/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Miócitos Cardíacos/metabolismo , Trifosfato de Adenosina/metabolismo , Canais KATP/genética , Canais KATP/metabolismo
3.
Physiol Rev ; 96(1): 177-252, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26660852

RESUMO

KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.


Assuntos
Doenças Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Canais KATP/metabolismo , Transdução de Sinais , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/fisiopatologia , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Canais KATP/antagonistas & inibidores , Canais KATP/química , Canais KATP/genética , Masculino , Potenciais da Membrana , Camundongos Transgênicos , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Conformação Proteica , Subunidades Proteicas , Transporte Proteico , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
4.
Proc Natl Acad Sci U S A ; 117(19): 10593-10602, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32332165

RESUMO

A physiological role for long-chain acyl-CoA esters to activate ATP-sensitive K+ (KATP) channels is well established. Circulating palmitate is transported into cells and converted to palmitoyl-CoA, which is a substrate for palmitoylation. We found that palmitoyl-CoA, but not palmitic acid, activated the channel when applied acutely. We have altered the palmitoylation state by preincubating cells with micromolar concentrations of palmitic acid or by inhibiting protein thioesterases. With acyl-biotin exchange assays we found that Kir6.2, but not sulfonylurea receptor (SUR)1 or SUR2, was palmitoylated. These interventions increased the KATP channel mean patch current, increased the open time, and decreased the apparent sensitivity to ATP without affecting surface expression. Similar data were obtained in transfected cells, rat insulin-secreting INS-1 cells, and isolated cardiac myocytes. Kir6.2ΔC36, expressed without SUR, was also positively regulated by palmitoylation. Mutagenesis of Kir6.2 Cys166 prevented these effects. Clinical variants in KCNJ11 that affect Cys166 had a similar gain-of-function phenotype, but was more pronounced. Molecular modeling studies suggested that palmitoyl-C166 and selected large hydrophobic mutations make direct hydrophobic contact with Kir6.2-bound PIP2 Patch-clamp studies confirmed that palmitoylation of Kir6.2 at Cys166 enhanced the PIP2 sensitivity of the channel. Physiological relevance is suggested since palmitoylation blunted the regulation of KATP channels by α1-adrenoreceptor stimulation. The Cys166 residue is conserved in some other Kir family members (Kir6.1 and Kir3, but not Kir2), which are also subject to regulated palmitoylation, suggesting a general mechanism to control the open state of certain Kir channels.


Assuntos
Canais KATP/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Acil Coenzima A/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cisteína/metabolismo , Células HEK293 , Humanos , Canais KATP/genética , Lipoilação/fisiologia , Mutagênese/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp/métodos , Canais de Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Cultura Primária de Células , Ratos , Receptores de Sulfonilureias/genética
5.
Am J Physiol Cell Physiol ; 322(6): C1230-C1247, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35508187

RESUMO

Sarcolemmal/plasmalemmal ATP-sensitive K+ (KATP) channels have key roles in many cell types and tissues. Hundreds of studies have described how the KATP channel activity and ATP sensitivity can be regulated by changes in the cellular metabolic state, by receptor signaling pathways and by pharmacological interventions. These alterations in channel activity directly translate to alterations in cell or tissue function, that can range from modulating secretory responses, such as insulin release from pancreatic ß-cells or neurotransmitters from neurons, to modulating contractile behavior of smooth muscle or cardiac cells to elicit alterations in blood flow or cardiac contractility. It is increasingly becoming apparent, however, that KATP channels are regulated beyond changes in their activity. Recent studies have highlighted that KATP channel surface expression is a tightly regulated process with similar implications in health and disease. The surface expression of KATP channels is finely balanced by several trafficking steps including synthesis, assembly, anterograde trafficking, membrane anchoring, endocytosis, endocytic recycling, and degradation. This review aims to summarize the physiological and pathophysiological implications of KATP channel trafficking and mechanisms that regulate KATP channel trafficking. A better understanding of this topic has potential to identify new approaches to develop therapeutically useful drugs to treat KATP channel-related diseases.


Assuntos
Células Secretoras de Insulina , Trifosfato de Adenosina/metabolismo , Transporte Biológico , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Canais KATP/genética , Canais KATP/metabolismo , Transporte Proteico
6.
J Mol Cell Cardiol ; 144: 1-11, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32339567

RESUMO

BACKGROUND: Genetic variants in SCN5A can result in channelopathies such as the long QT syndrome type 3 (LQT3), but the therapeutic response to Na+ channel blockers can vary. We previously reported a case of an infant with malignant LQT3 and a missense Q1475P SCN5A variant, who was effectively treated with phenytoin, but only partially with mexiletine. Here, we functionally characterized this variant and investigated possible mechanisms for the differential drug actions. METHODS: Wild-type or mutant Nav1.5 cDNAs were examined in transfected HEK293 cells with patch clamping and biochemical assays. We used computational modeling to provide insights into altered channel kinetics and to predict effects on the action potential. RESULTS: The Q1475P variant in Nav1.5 reduced the current density and channel surface expression, characteristic of a trafficking defect. The variant also led to positive shifts in the voltage dependence of steady-state activation and inactivation, faster inactivation and recovery from inactivation, and increased the "late" Na+ current. Simulations of Nav1.5 gating with a 9-state Markov model suggested that transitions from inactivated to closed states were accelerated in Q1475P channels, leading to accumulation of channels in non-inactivated closed states. Simulations with a human ventricular myocyte model predicted action potential prolongation with Q1475P, compared with wild type, channels. Patch clamp data showed that mexiletine and phenytoin similarly rescued some of the gating defects. Chronic incubation with mexiletine, but not phenytoin, rescued the Nav1.5-Q1475P trafficking defect, thus increasing mutant channel expression. CONCLUSIONS: The gain-of-function effects of Nav1.5-Q1475P predominate to cause a malignant long QT phenotype. Phenytoin partially corrects the gating defect without restoring surface expression of the mutant channel, whereas mexiletine restores surface expression of the mutant channel, which may explain the lack of efficacy of mexiletine when compared to phenytoin. Our data makes a case for experimental studies before embarking on a one-for-all therapy of arrhythmias.


Assuntos
Doença do Sistema de Condução Cardíaco/etiologia , Gerenciamento Clínico , Suscetibilidade a Doenças , Síndrome do QT Longo/etiologia , Fenitoína/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/uso terapêutico , Potenciais de Ação/efeitos dos fármacos , Substituição de Aminoácidos , Antiarrítmicos/farmacologia , Doença do Sistema de Condução Cardíaco/diagnóstico , Doença do Sistema de Condução Cardíaco/tratamento farmacológico , Doença do Sistema de Condução Cardíaco/metabolismo , Células Cultivadas , Mutação com Ganho de Função , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/metabolismo , Modelos Biológicos , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Técnicas de Patch-Clamp , Fenitoína/uso terapêutico
7.
FASEB J ; 32(3): 1613-1625, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29133341

RESUMO

ATP-sensitive K+ (KATP) channels uniquely link cellular energy metabolism to membrane excitability and are expressed in diverse cell types that range from the endocrine pancreas to neurons and smooth, skeletal, and cardiac muscle. A decrease in the surface expression of KATP channels has been linked to various disorders, including dysregulated insulin secretion, abnormal blood pressure, and impaired resistance to cardiac injury. In contrast, up-regulation of KATP channel surface expression may be protective, for example, by mediating the beneficial effect of ischemic preconditioning. Molecular mechanisms that regulate KATP channel trafficking are poorly understood. Here, we used cellular assays with immunofluorescence, surface biotinylation, and patch clamping to demonstrate that Eps15 homology domain-containing protein 2 (EHD2) is a novel positive regulator of KATP channel trafficking to increase surface KATP channel density. EHD2 had no effect on cardiac Na+ channels (Nav1.5). The effect is specific to EHD2 as other members of the EHD family-EHD1, EHD3, and EHD4-had no effect on KATP channel surface expression. EHD2 did not directly affect KATP channel properties as unitary conductance and ATP sensitivity were unchanged. Instead, we observed that the mechanism by which EHD2 increases surface expression is by stabilizing KATP channel-containing caveolar structures, which results in a reduced rate of endocytosis. EHD2 also regulated KATP channel trafficking in isolated cardiomyocytes, which validated the physiologic relevance of these observations. Pathophysiologically, EHD2 may be cardioprotective as a dominant-negative EHD2 mutant sensitized cardiomyocytes to ischemic damage. Our findings highlight EHD2 as a potential pharmacologic target in the treatment of diseases with KATP channel trafficking defects.-Yang, H. Q., Jana, K., Rindler, M. J., Coetzee, W. A. The trafficking protein, EHD2, positively regulates cardiac sarcolemmal KATP channel surface expression: role in cardioprotection.


Assuntos
Proteínas de Transporte/metabolismo , Canais KATP/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Sarcolema/metabolismo , Animais , Células COS , Proteínas de Transporte/genética , Chlorocebus aethiops , Células HEK293 , Humanos , Canais KATP/genética , Camundongos , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Transporte Proteico , Ratos , Sarcolema/genética
8.
Pacing Clin Electrophysiol ; 42(2): 275-282, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30578647

RESUMO

The HCN4 gene encodes a subunit of the hyperpolarization-activated cyclic nucleotide-gated channel, type 4 that is essential for the proper generation of pacemaker potentials in the sinoatrial node. The HCN4 gene is often present in targeted genetic testing panels for various cardiac conduction system disorders and there are several reports of HCN4 variants associated with conduction disorders. Here, we report the in vitro functional characterization of four rare variants of uncertain significance (VUS) in HCN4, identified through testing a cohort of 296 sudden unexpected natural deaths. The variants are all missense alterations, leading to single amino acid changes: p.E66Q in the N-terminus, p.D546N in the C-linker domain, and both p.S935Y and p.R1044Q in the C-terminus distal to the CNBD. We also identified a likely benign variant, p. P1063T, which has a high minor allele frequency in the gnomAD, which is utilized here as a negative control. Three of the HCN4 VUS (p.E66Q, p.S935Y, and p.R1044Q) had electrophysiological characteristics similar to the wild-type channel, suggesting that these variants are benign. In contrast, the p.D546N variant in the C-linker domain exhibited a larger current density, slower activation, and was unresponsive to cyclic adenosine monophosphate (cAMP) compared to wild-type. With functional assays, we reclassified three rare HCN4 VUS to likely benign variants, eliminating the necessity for costly and time-consuming further study. Our studies also provide a new lead to investigate how a VUS located in the C-linker connecting the pore to the cAMP binding domain may affect the channel open state probability and cAMP response.


Assuntos
Morte Súbita Cardíaca , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/classificação , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Proteínas Musculares/classificação , Proteínas Musculares/genética , Canais de Potássio/classificação , Canais de Potássio/genética , Células Cultivadas , Fenômenos Eletrofisiológicos , Variação Genética , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/fisiologia , Proteínas Musculares/fisiologia , Canais de Potássio/fisiologia
9.
Forensic Sci Med Pathol ; 15(3): 437-444, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30547356

RESUMO

Determining the cause of unexplained death in all age groups, including infants, is a priority in forensic medicine. The triple risk model proposed for sudden infant death syndrome involves the intersection of three risks: (1) a critical developmental period in homeostatic control (2), exogenous stressors, and (3) a vulnerable infant. Even though sex and age factor into some forms of inherited arrhythmogenic deaths in young individuals and adults, more appropriate a dual-risk disease model for adults involves exogenous stressors and a vulnerable individual. The vulnerability aspect clearly has a genetic component as underscored by a number of recent large-scale and high-throughput genetic testing studies performed in attempt to define the causes of sudden unexplained death. These studies often focus on 'cardiac' and channelopathy genes. Genetic testing often identify lists of rare or ultra-rare nonsynonymous variants, classified according to the ACMG guidelines as 'pathogenic' or 'likely pathogenic', which may form the basis of diagnostic decisions and/or family counseling. However, computer algorithms used to categorize gene variants are not completely accurate and these variants are often not functionally tested to determine their pathogenicity. Due to conflicting computational predictions, a large number of variants are labeled as 'variants of uncertain significance' or VUS. Functional testing of these VUS can greatly assist to reclassify these VUS as 'likely benign' or 'likely pathogenic'. However, functional testing has its limits and by itself cannot be used to determine cause of death. Going forward, computer algorithms must be improved to take account of variants across multiple genes and efforts must be expanded to obtain clinical, familial and segregation data. Forensic genetic testing needs to be held to the same rigorous standards as defined by the NIH Clinical Genome Resource Consortium, where functional evaluation of a channelopathy variant is only one (but important) aspect of the overall picture.


Assuntos
Canalopatias/genética , Morte Súbita/etiologia , Variação Genética , Morte Súbita do Lactente/genética , Genética Forense , Predisposição Genética para Doença , Testes Genéticos , Humanos , Lactente
10.
Artigo em Inglês | MEDLINE | ID: mdl-29781517

RESUMO

BACKGROUND: Atrial standstill is an arrhythmogenic condition characterized by the absence of spontaneous electrical and mechanical atrial activity or in response to stimulation. There are few reported familial cases which have been associated with SCN5A mutations cosegregating with GJA5 or RYR2; however, isolated SCN5A mutations are rare. OBJECTIVE: The purpose of this study was to determine the clinical and biophysical consequence of a novel SCN5A mutation identified in a family with progressive atrial standstill and sudden death. METHODS: The family of a sporadic case of congenital atrial standstill underwent genetic screening. Human Embryonic Kidney 293 cells were transfected with wild-type (WT) or mutant SCN5A cDNAs. Biophysical properties were studied using whole-cell using patch clamp methods. RESULTS: A novel homozygous SCN5A mutation, p.V1340L, was identified in the proband and her sister. The proband had complete atrial standstill whereas the sister had partial atrial standstill. Heterozygous mutations were identified in the mother, father, and brother. All three had normal sinus rhythm and were asymptomatic. The mutant Nav1.5(V1340L) reduced Nav1.5 current density as well as showed a depolarizing shift in the voltage-dependent steady-state activation (WT: -35.3 ± 1.62 mV; V1340L: -22.4 ± 2.59 mV; P  =  0.001). CONCLUSIONS: A homozygous loss-of-function SCN5A mutation likely results in atrial standstill and sudden death due to suppression of initiation of action potential.

11.
Pacing Clin Electrophysiol ; 40(6): 703-712, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28370132

RESUMO

BACKGROUND: Two genetic variants in SCN5A, encoding the Nav1.5 Na+ channel α-subunit, were found in a 5-month-old girl who died suddenly in her sleep. The first variant is a missense mutation, resulting in an amino acid change (Q1832E), which has been described (but not characterized) in a patient with Brugada syndrome. The second is a nonsense mutation that produces a premature stop codon and a C-terminal truncation (R1944Δ). METHODS AND RESULTS: To investigate their functional relevance with patch clamp experiments in transfected HEK-293 cells. The Q1832E mutation drastically reduced Nav1.5 current density. The R1944Δ C-terminal truncation had negligible effects on Nav1.5 current density. Neither of the mutations affected the voltage dependence of steady activation and inactivation or influenced the late Na+ current or the recovery from inactivation. Biochemical and immunofluorescent approaches demonstrated that the Q1832E mutation caused severe trafficking defects. Polymerase chain reaction cloning and sequencing the victim's genomic DNA allowed us to determine that the two variants were in trans. We investigated the functional consequences by coexpressing Nav1.5(Q1832E) and Nav1.5(R1944Δ), which led to a significantly reduced current amplitude relative to wild-type. CONCLUSIONS: These sudden infant death syndrome (SIDS)-related variants caused a severely dysfunctional Nav1.5 channel, which was mainly due to trafficking defects caused by the Q1832E mutation. The decreased current density is likely to be a major contributing factor to arrhythmogenesis in Brugada syndrome and the sudden death of this SIDS victim.


Assuntos
Ativação do Canal Iônico/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Sódio/metabolismo , Morte Súbita do Lactente/genética , Morte Súbita Cardíaca , Feminino , Marcadores Genéticos/genética , Predisposição Genética para Doença/genética , Células HEK293 , Humanos , Incidência , Lactente , Mutação/genética , Fatores de Risco , Taxa de Sobrevida
12.
Am J Physiol Heart Circ Physiol ; 310(11): H1558-66, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27037371

RESUMO

Myocardial ischemia remains the primary cause of morbidity and mortality in the United States. Ischemic preconditioning (IPC) is a powerful form of endogenous protection against myocardial infarction. We studied alterations in KATP channels surface density as a potential mechanism of the protection of IPC. Using cardiac-specific knockout of Kir6.2 subunits, we demonstrated an essential role for sarcolemmal KATP channels in the infarct-limiting effect of IPC in the mouse heart. With biochemical membrane fractionation, we demonstrated that sarcolemmal KATP channel subunits are distributed both to the sarcolemma and intracellular endosomal compartments. Global ischemia causes a loss of sarcolemmal KATP channel subunit distribution and internalization to endosomal compartments. Ischemia-induced internalization of KATP channels was prevented by CaMKII inhibition. KATP channel subcellular redistribution was also observed with immunohistochemistry. Ischemic preconditioning before the index ischemia reduced not only the infarct size but also prevented KATP channel internalization. Furthermore, not only did adenosine mimic IPC by preventing infarct size, but it also prevented ischemia-induced KATP channel internalization via a PKC-mediated pathway. We show that preventing endocytosis with dynasore reduced both KATP channel internalization and strongly mitigated infarct development. Our data demonstrate that plasticity of KATP channel surface expression must be considered as a potentially important mechanism of the protective effects of IPC and adenosine.


Assuntos
Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Sarcolema/metabolismo , Adenosina/farmacologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Endocitose , Endossomos/metabolismo , Hidrazonas/farmacologia , Preparação de Coração Isolado , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/deficiência , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Sarcolema/efeitos dos fármacos , Fatores de Tempo
13.
Biochem Biophys Res Commun ; 471(1): 129-34, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26828268

RESUMO

Sarcolemmal ATP-sensitive potassium (KATP) channels control skeletal muscle energy use through their ability to adjust membrane excitability and related cell functions in accordance with cellular metabolic status. Mice with disrupted skeletal muscle KATP channels exhibit reduced adipocyte size and increased fatty acid release into the circulation. As yet, the molecular mechanisms underlying this link between skeletal muscle KATP channel function and adipose mobilization have not been established. Here, we demonstrate that skeletal muscle-specific disruption of KATP channel function in transgenic (TG) mice promotes production and secretion of musclin. Musclin is a myokine with high homology to atrial natriuretic peptide (ANP) that enhances ANP signaling by competing for elimination. Augmented musclin production in TG mice is driven by a molecular cascade resulting in enhanced acetylation and nuclear exclusion of the transcription factor forkhead box O1 (FOXO1) - an inhibitor of transcription of the musclin encoding gene. Musclin production/secretion in TG is paired with increased mobilization of fatty acids and a clear trend toward increased circulating ANP, an activator of lipolysis. These data establish KATP channel-dependent musclin production as a potential mechanistic link coupling "local" skeletal muscle energy consumption with mobilization of bodily resources from fat. Understanding such mechanisms is an important step toward designing interventions to manage metabolic disorders including those related to excess body fat and associated co-morbidities.


Assuntos
Ativação do Canal Iônico/fisiologia , Canais KATP/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Fatores de Transcrição/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos
14.
J Physiol ; 593(6): 1389-407, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25772295

RESUMO

KEY POINTS: Na(+) current (INa) results from the integrated function of a molecular aggregate (the voltage-gated Na(+) channel complex) that includes the ß subunit family. Mutations or rare variants in Scn1b (encoding the ß1 and ß1B subunits) have been associated with various inherited arrhythmogenic syndromes, including Brugada syndrome and sudden unexpected death in patients with epilepsy. We used Scn1b null mice to understand better the relation between Scn1b expression, and cardiac electrical function. Loss of Scn1b caused, among other effects, increased amplitude of tetrodotoxin-sensitive INa, delayed after-depolarizations, triggered beats, delayed Ca(2+) transients, frequent spontaneous calcium release events and increased susceptibility to polymorphic ventricular arrhythmias. Most alterations in Ca(2+) homeostasis were prevented by 100 nM tetrodotoxin. We propose that life-threatening arrhythmias in patients with mutations in Scn1b, a gene classically defined as ancillary to the Na(+) channel α subunit, can be partly consequent to disrupted intracellular Ca(2+) homeostasis. ABSTRACT: Na(+) current (INa) is determined not only by the properties of the pore-forming voltage-gated Na(+) channel (VGSC) α subunit, but also by the integrated function of a molecular aggregate (the VGSC complex) that includes the VGSC ß subunit family. Mutations or rare variants in Scn1b (encoding the ß1 and ß1B subunits) have been associated with various inherited arrhythmogenic syndromes, including cases of Brugada syndrome and sudden unexpected death in patients with epilepsy. Here, we have used Scn1b null mouse models to understand better the relation between Scn1b expression, and cardiac electrical function. Using a combination of macropatch and scanning ion conductance microscopy we show that loss of Scn1b in juvenile null animals resulted in increased tetrodotoxin-sensitive INa but only in the cell midsection, even before full T-tubule formation; the latter occurred concurrent with increased message abundance for the neuronal Scn3a mRNA, suggesting increased abundance of tetrodotoxin-sensitive NaV 1.3 protein and yet its exclusion from the region of the intercalated disc. Ventricular myocytes from cardiac-specific adult Scn1b null animals showed increased Scn3a message, prolonged action potential repolarization, presence of delayed after-depolarizations and triggered beats, delayed Ca(2+) transients and frequent spontaneous Ca(2+) release events and at the whole heart level, increased susceptibility to polymorphic ventricular arrhythmias. Most alterations in Ca(2+) homeostasis were prevented by 100 nM tetrodotoxin. Our results suggest that life-threatening arrhythmias in patients with mutations in Scn1b, a gene classically defined as ancillary to the Na(+) channel α subunit, can be partly consequent to disrupted intracellular Ca(2+) homeostasis in ventricular myocytes.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/genética , Sinalização do Cálcio , Miócitos Cardíacos/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem/genética , Animais , Arritmias Cardíacas/metabolismo , Células Cultivadas , Deleção de Genes , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(3): E154-63, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22203979

RESUMO

TBX3 is critical for human development: mutations in TBX3 cause congenital anomalies in patients with ulnar-mammary syndrome. Data from mice and humans suggest multiple roles for Tbx3 in development and function of the cardiac conduction system. The mechanisms underlying the functional development, maturation, and maintenance of the conduction system are not well understood. We tested the requirements for Tbx3 in these processes. We generated a unique series of Tbx3 hypomorphic and conditional mouse mutants with varying levels and locations of Tbx3 activity within the heart, and developed techniques for evaluating in vivo embryonic conduction system function. Disruption of Tbx3 function in different regions of the developing heart causes discrete phenotypes and lethal arrhythmias: sinus pauses and bradycardia indicate sinoatrial node dysfunction, whereas preexcitation and atrioventricular block reveal abnormalities in the atrioventricular junction. Surviving Tbx3 mutants are at increased risk for sudden death. Arrhythmias induced by knockdown of Tbx3 in adults reveal its requirement for conduction system homeostasis. Arrhythmias in Tbx3-deficient embryos are accompanied by disrupted expression of multiple ion channels despite preserved expression of previously described conduction system markers. These findings indicate that Tbx3 is required for the conduction system to establish and maintain its correct molecular identity and functional properties. In conclusion, Tbx3 is required for the functional development, maturation, and homeostasis of the conduction system in a highly dosage-sensitive manner. TBX3 and its regulatory targets merit investigation as candidates for human arrhythmias.


Assuntos
Arritmias Cardíacas/fisiopatologia , Dosagem de Genes , Sistema de Condução Cardíaco/fisiopatologia , Homeostase/genética , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Alelos , Animais , Animais Recém-Nascidos , Arritmias Cardíacas/complicações , Arritmias Cardíacas/diagnóstico por imagem , Arritmias Cardíacas/patologia , Bloqueio Atrioventricular/complicações , Bloqueio Atrioventricular/diagnóstico por imagem , Bloqueio Atrioventricular/patologia , Bloqueio Atrioventricular/fisiopatologia , Nó Atrioventricular/patologia , Nó Atrioventricular/fisiopatologia , Conexina 43/metabolismo , Eletrocardiografia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Sistema de Condução Cardíaco/anormalidades , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/patologia , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Mutação/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética/genética , Análise de Sobrevida , Proteínas com Domínio T/metabolismo , Ultrassonografia
16.
bioRxiv ; 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39211194

RESUMO

Involved in immunity and reproduction, natural killer (NK) cells offer opportunities to develop new immunotherapies to treat infections and cancer or to alleviate pregnancy complications. Most current strategies use cytokines or antibodies to enhance NK-cell function, but none use ion channel modulators, which are widely used in clinical practice to treat hypertension, diabetes, epilepsy, and other conditions. Little is known about ion channels in NK cells. We show that Kcnj8, which codes for the Kir6.1 subunit of a certain type of ATP-sensitive potassium (K ATP ) channel, is highly expressed in murine splenic and uterine NK cells compared to other K + channels previously identified in NK cells. Kcnj8 expression is highest in the most mature subset of splenic NK cells (CD27 - CD11b + ) and in NKG2A + or Ly49C/I + educated uterine NK cells. Using patch clamping, we show that a subset of NK cells expresses a current sensitive to the Kir6.1 blocker PNU-37883A. Kcnj8 does not participate in NK cell degranulation in response to tumor cells in vitro or rejection of tumor cells in vivo . Transcriptomics show that genes previously implicated in NK cell development are amongst those differentially expressed in CD27 - CD11b + NK cells deficient of Kcnj8 . Indeed, we found that mice with NK-cell specific Kcnj8 gene ablation have fewer CD11b + CD27 - and KLRG-1 + NK cells in the bone barrow and spleen. These results show that the K ATP subunit Kir6.1 has a key role in NK-cell development.

17.
Proteomics ; 13(2): 368-78, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23197389

RESUMO

ATP-sensitive K(+) (K(ATP)) channels are expressed ubiquitously, but have diverse roles in various organs and cells. Their diversity can partly be explained by distinct tissue-specific compositions of four copies of the pore-forming inward rectifier potassium channel subunits (Kir6.1 and/or Kir6.2) and four regulatory sulfonylurea receptor subunits (SUR1 and/or SUR2). Channel function and/or subcellular localization also can be modified by the proteins with which they transiently or permanently interact to generate even more diversity. We performed a quantitative proteomic analysis of K(ATP) channel complexes in the heart, endothelium, insulin-secreting min6 cells (pancreatic ß-cell like), and the hypothalamus to identify proteins with which they interact in different tissues. Glycolysis is an overrepresented pathway in identified proteins of the heart, min6 cells, and the endothelium. Proteins with other energy metabolic functions were identified in the hypothalamic samples. These data suggest that the metabolo-electrical coupling conferred by K(ATP) channels is conferred partly by proteins with which they interact. A large number of identified cytoskeletal and trafficking proteins suggests endocytic recycling may help control K(ATP) channel surface density and/or subcellular localization. Overall, our data demonstrate that K(ATP) channels in different tissues may assemble with proteins having common functions, but that tissue-specific complex organization also occurs.


Assuntos
Canais KATP/química , Canais KATP/metabolismo , Proteômica/métodos , Transportadores de Cassetes de Ligação de ATP , Animais , Endotélio/química , Endotélio/metabolismo , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Canais KATP/análise , Camundongos , Miocárdio/química , Miocárdio/metabolismo , Especificidade de Órgãos , Canais de Potássio Corretores do Fluxo de Internalização , Receptores de Droga , Receptores de Sulfonilureias
18.
J Biol Chem ; 287(49): 41258-67, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23066018

RESUMO

Ventricular ATP-sensitive potassium (K(ATP)) channels link intracellular energy metabolism to membrane excitability and contractility. Our recent proteomics experiments identified plakoglobin and plakophilin-2 (PKP2) as putative K(ATP) channel-associated proteins. We investigated whether the association of K(ATP) channel subunits with junctional proteins translates to heterogeneous subcellular distribution within a cardiac myocyte. Co-immunoprecipitation experiments confirmed physical interaction between K(ATP) channels and PKP2 and plakoglobin in rat heart. Immunolocalization experiments demonstrated that K(ATP) channel subunits (Kir6.2 and SUR2A) are expressed at a higher density at the intercalated disk in mouse and rat hearts, where they co-localized with PKP2 and plakoglobin. Super-resolution microscopy demonstrate that K(ATP) channels are clustered within nanometer distances from junctional proteins. The local K(ATP) channel density, recorded in excised inside-out patches, was larger at the cell end when compared with local currents recorded from the cell center. The K(ATP) channel unitary conductance, block by MgATP and activation by MgADP, did not differ between these two locations. Whole cell K(ATP) channel current density (activated by metabolic inhibition) was ∼40% smaller in myocytes from mice haploinsufficient for PKP2. Experiments with excised patches demonstrated that the regional heterogeneity of K(ATP) channels was absent in the PKP2 deficient mice, but the K(ATP) channel unitary conductance and nucleotide sensitivities remained unaltered. Our data demonstrate heterogeneity of K(ATP) channel distribution within a cardiac myocyte. The higher K(ATP) channel density at the intercalated disk implies a possible role at the intercellular junctions during cardiac ischemia.


Assuntos
Canais KATP/química , Miócitos Cardíacos/citologia , Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Animais , Membrana Celular/metabolismo , Desmossomos/metabolismo , Masculino , Camundongos , Microscopia/métodos , Isquemia Miocárdica/patologia , Placofilinas/metabolismo , Ratos , Ratos Sprague-Dawley , gama Catenina/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 304(9): H1231-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23436329

RESUMO

Cardiac metabolism remains altered for an extended period of time after myocardial infarction. Studies have shown fibroblasts from normal hearts express KATP channels in culture. It is unknown whether fibroblasts from infarcted hearts express KATP channels and whether these channels contribute to scar and border zone electrophysiology. KATP channel subunit expression levels were determined in fibroblasts isolated from normal hearts (Fb), and scar (sMI-Fb) and remote (rMI-Fb) regions of left anterior descending coronary artery (LAD) ligated rat hearts. Whole cell KATP current density was determined with patch clamp. Action potential duration (APD) was measured with optical mapping in myocyte-only cultures and heterocellular cultures with fibroblasts with and without 100 µmol/l pinacidil. Whole heart optical mapping was used to assess KATP channel activity following LAD ligation. Pinacidil activated a potassium current (35.4 ± 7.5 pA/pF at 50 mV) in sMI-Fb that was inhibited with 10 µmol/l glibenclamide. Kir6.2 and SUR2 transcript levels were elevated in sMI-Fb. Treatment with Kir6.2 short interfering RNA decreased KATP currents (87%) in sMI-Fb. Treatment with pinacidil decreased APD (26%) in co-cultures with sMI-Fb. APD values were prolonged in LAD ligated hearts after perfusion with glibenclamide. KATP channels are present in fibroblasts from the scar and border zones of infarcted hearts. Activation of fibroblast KATP channels could modulate the electrophysiological substrate beyond the acute ischemic event. Targeting fibroblast KATP channels could represent a novel therapeutic approach to modify border zone electrophysiology after cardiac injury.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Fibroblastos/fisiologia , Canais KATP/fisiologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/fisiologia , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Potenciais de Ação/fisiologia , Animais , Fibroblastos/metabolismo , Glibureto/farmacologia , Ventrículos do Coração/citologia , Canais KATP/agonistas , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Masculino , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Pinacidil/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Ratos , Ratos Wistar , Receptores de Droga/genética , Receptores de Droga/metabolismo , Receptores de Sulfonilureias , Transcrição Gênica , Imagens com Corantes Sensíveis à Voltagem
20.
Circ Res ; 109(3): 291-5, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21680895

RESUMO

RATIONALE: Flecainide prevents arrhythmias in catecholaminergic polymorphic ventricular tachycardia, but the antiarrhythmic mechanism remains unresolved. It is possible for flecainide to directly affect the cardiac ryanodine receptor (RyR2); however, an extracellular site of action is suggested because of the hydrophilic nature of flecainide. OBJECTIVE: To investigate the mechanism for the antiarrhythmic action of flecainide in a RyR2(R4496C+/-) knock-in mouse model of catecholaminergic polymorphic ventricular tachycardia. METHODS AND RESULTS: Flecainide prevented catecholamine-induced sustained ventricular tachycardia in RyR2(R4496C+/-) mice. Cellular studies were performed with isolated RyR2(R4496C+/-) myocytes. Isoproterenol caused the appearance of spontaneous Ca(2+) transients, which were unaffected by flecainide (6 µmol/L). Flecainide did not affect Ca(2+) transient amplitude, decay, or sarcoplasmic reticulum Ca(2+) content. Moreover, it did not affect the frequency of spontaneous Ca(2+) sparks in permeabilized myocytes. In contrast, flecainide effectively prevented triggered activity induced by isoproterenol. The threshold for action potential induction was increased significantly (P<0.01), which suggests a primary extracellular antiarrhythmic effect mediated by Na(+) channel blockade. CONCLUSIONS: Flecainide prevents catecholaminergic polymorphic ventricular tachycardia in RyR2(R4496C+/-) mice; however, at variance with previous reports, we observed minimal effects on intracellular Ca(2+) homeostasis. Our data suggest that the antiarrhythmic activity of the drug is caused by reduction of Na(+) channel availability and by an increase in the threshold for triggered activity.


Assuntos
Antiarrítmicos/farmacologia , Flecainida/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Taquicardia Ventricular/tratamento farmacológico , Taquicardia Ventricular/prevenção & controle , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Modelos Animais de Doenças , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/fisiologia , Técnicas de Introdução de Genes , Isoproterenol/farmacologia , Camundongos , Camundongos Mutantes , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/fisiologia , Canais de Sódio/fisiologia , Simpatomiméticos/farmacologia , Taquicardia Ventricular/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa